EP2585071A1 - Composition pharmaceutique destinée à être utilisée dans le traitement du glaucome - Google Patents

Composition pharmaceutique destinée à être utilisée dans le traitement du glaucome

Info

Publication number
EP2585071A1
EP2585071A1 EP11727194.0A EP11727194A EP2585071A1 EP 2585071 A1 EP2585071 A1 EP 2585071A1 EP 11727194 A EP11727194 A EP 11727194A EP 2585071 A1 EP2585071 A1 EP 2585071A1
Authority
EP
European Patent Office
Prior art keywords
cxcr3
glaucoma
compound according
see
iop
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP11727194.0A
Other languages
German (de)
English (en)
Inventor
William Rostene
Alexandre Denoyer
Christophe Baudouin
David Godefroy
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Institut National de la Sante et de la Recherche Medicale INSERM
Original Assignee
Institut National de la Sante et de la Recherche Medicale INSERM
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Institut National de la Sante et de la Recherche Medicale INSERM filed Critical Institut National de la Sante et de la Recherche Medicale INSERM
Priority to EP11727194.0A priority Critical patent/EP2585071A1/fr
Publication of EP2585071A1 publication Critical patent/EP2585071A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the invention relates to a compound which is an antagonist of CXCR3 or an inhibitor of the CXCR3 receptor expression for use in the treatment of glaucoma.
  • Glaucoma is one of the most frequent ocular diseases which prevalence averages about 2% which is increasing with the aging of the population. Despite of the wide advances in the medical and surgical managements of glaucoma, it still leads to clinical blindness in 1% to 30% of the patients, depending on the racial group and the country. So, there is a permanent need in the art for new molecules for the treatment of glaucoma (see for example Cedrone C. et AL, 2008). SUMMARY OF THE INVENTION
  • Trabecular meshwork is a functional and anatomic ocular entity located at the angle formed by the cornea and iris, which primary function is to control the intraocular pressure (IOP) by regulating the aqueous humor (AH) outflow.
  • IOP intraocular pressure
  • AH aqueous humor
  • a first object of the invention relates to a compound which is an antagonist of
  • CXCR3 or an inhibitor of the CXCR3 receptor expression for use in the treatment of glaucoma.
  • Another object of the invention relates to a pharmaceutical composition for the treatment of glaucoma comprising a compound according to the invention and a pharmaceutically acceptable carrier.
  • CXCR3 is well known.
  • CXCR3 is a God protein-coupled receptor in the CXC chemokine receptor family.
  • Other names for CXCR3 are G protein-coupled receptor 9 (GPR9) and CD 183.
  • GPR9 G protein-coupled receptor 9
  • CXCR3-A binds to the CXC chemokines CXCL9 (MIG), CXCL10 (IP-10), and CXCL1 1 (I-TAC)
  • CXCR3-B can also bind to CXCL4 in addition to CXCL9, CXCL 10, and CXCL1 1.
  • CXCR3 is able to regulate leukocyte trafficking.
  • CXCR3 Binding of chemokines to CXCR3 induces various cellular responses, most notably integrin activation, cytoskeletal changes and chemotactic migration. CXCR3-ligand interaction attracts Thl cells and promotes Thl cell maturation. CXCR3 is well known. As explained in the article Pease et al, (Pease et al, 2009, Expert Opin. Ther. Patents, 19(l):39-58) there are evidence for a role of CXCR3 in the pathophysiology of autoimmune diseases like psoriasis, transplant rejection and T-cell trafficking to sites of inflammation.
  • treating denotes reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or reversing, alleviating, inhibiting the progress of, or preventing one or more symptoms of the disorder or condition to which such term applies.
  • Glaucoma may be divided in primary and secondary glaucoma.
  • Primary glaucoma may be divided in "open angle” and “angle-closure” glaucoma.
  • the compound according to the invention may be used to primary or secondary glaucoma.
  • a first object of the invention relates to a compound which is an antagonist of CXCR3 or an inhibitor of the CXCR3 receptor expression for use in the treatment of glaucoma.
  • said compound according to the invention is a CXCR3 antagonist.
  • said CXCR3 antagonist may be a low molecular weight antagonist, e. g. a small organic molecule (natural or not).
  • small organic molecule refers to a molecule (natural or not) of a size comparable to those organic molecules generally used in pharmaceuticals.
  • Preferred small organic molecules range in size up to about 10000 Da, more preferably up to 5000 Da, more preferably up to 2000 Da and most preferably up to about 1000 Da.
  • the antagonist may bind to CXCR3 and block the binding of other compound on CXCR3.
  • the compound according to the invention is NBI74330 (see for example Jopling LA et AL, 2007), AMG487 (see for example Jiwen Liu, et al, 2009, An-Rong Lia et al, 2008 or Johnson M. et al, 2007), AMG1237845 (see for example Rosenblum JM et al, 2009).
  • Antagonists of CXCR3 are well known in the state of the art (see for example, Pease J.E. el al, 2009, Expert Opin. Ther. Patents).
  • the compound according to the invention may be a dihydro-quinazoline analog (see for example Pease J.E. el al, 2009, Expert Opin. Ther. or Liu J et al, 2009) like AMG487 (see for example Jiwen Liu, et al, 2009, An-Rong Lia et al, 2008 or Johnson M. et al, 2007).
  • the compound according to the invention may be a piperidinyl-urea derivative (see for example Pease J.E. el al, 2009, Expert Opin. Ther.) like a l-aryl-3-piperidin-4-yl-urea derivative (see for example Allen et AL, 2007) or a 5-(piperidin- 4-yl)amino-l,2,4-thiadiazole derivative (see for example Watson et AL, 2007), or a tropenyl derivative (see for example Watson et AL, 2008) or a 2-aminoquinoline substituted piperidines derivative (see for example Knight et AL, 2008).
  • a piperidinyl-urea derivative see for example Pease J.E. el al, 2009, Expert Opin. Ther.
  • a piperidinyl-urea derivative see for example Pease J.E. el al, 2009, Expert Opin. Ther.
  • a piperidinyl-urea derivative see for example Pease J
  • the compound according to the invention may be a 4-aryl- [l,4]diazepine ethyl ureas derivative (see for example Pease J.E. el al, 2009, Expert Opin. Ther. and Cole AG, et al., 2006).
  • the compound according to the invention may be a benzimidazole derivative or a 2-iminobenzimidazole (see for example Pease J.E. el al, 2009, Expert Opin. Ther., Hayes ME, Wallace GA, et AL, 2008 and Hayes ME, Breinlinger EC et AL, 2008).
  • the compound according to the invention may be a benzetimide derivative (see for example Pease J.E. el al, 2009, Expert Opin. Ther. and Bongartz JP et al., 2008).
  • the compound according to the invention may be benzetimide derivative (see for example Pease J.E. el al, 2009, Expert Opin. Ther. and Bongartz JP et al., 2008).
  • the compound according to the invention may be an ergo line derivative (see for example Thoma G. et AL, 2009 or Choudhary MS et AL, 1995 and patent application WO2006128658).
  • the compound according to the invention may be a heterocyclic substituted piperazine derivative (see for example patent applications WO2006088837 and WO2008008453).
  • the compound according to the invention may be a heterocyclic substituted pyridine derivative (see for example patent applications WO2007109238 and WO2006088840).
  • the compound according to the invention may be a imidazolium derivative (see for example Verzijl D. Et al. , and patent application WO03101970).
  • the compound according to the invention may be an imidazole derivative (see for example Du X. et Al, 2008).
  • the compound according to the invention may be a piperazinyl-piperidine derivative (see for example McGuinness BF et AL, 2009).
  • the compound according to the invention may be a Camphor sulfonamide derivative (see for example Wang Y et AL, 2009).
  • the compound according to the invention may be a pyrazinyl substituted piperazine-piperidine derivative (see for example patent application WO2006088921).
  • the compound according to the invention may be a pyridyl and phenyl substituted piperazine-piperidine derivative (see for example patent application WO2006088919).
  • the compound according to the invention may be a piperazine-piperidine derivative (see for example patent application WO2006088836).
  • the compound according to the invention may be a heteroaryl substituted pyrazinyl-piperazine-piperidine derivative (see for example patent application WO2006091428).
  • the compound according to the invention may be an amine-linked pyridyl and phenyl substituted piperazine-piperidine derivative (see for example patent application WO2006088920).
  • the compound according to the invention may be a thiazole derivative (see for example patent application WO2007064553).
  • the compound according to the invention may be a substituted heterocyclic derivative (see for example patent application WO2007047202).
  • the compound according to the invention me be a compound described in Crosignani et al 2010.
  • CXCR3 antagonist of the invention may be an anti-CXCR3 antibody which neutralizes CXCR3 or an anti-CXCR3 fragment thereof which neutralizes CXCR3 (see for example Xie JH et AL, 2003).
  • Antibodies directed against CXCR3 can be raised according to known methods by administering the appropriate antigen or epitope to a host animal selected, e.g., from pigs, cows, horses, rabbits, goats, sheep, and mice, among others.
  • Various adjuvants known in the art can be used to enhance antibody production.
  • antibodies useful in practicing the invention can be polyclonal, monoclonal antibodies are preferred.
  • Monoclonal antibodies against CXCR3 can be prepared and isolated using any technique that provides for the production of antibody molecules by continuous cell lines in culture. Techniques for production and isolation include but are not limited to the hybridoma technique originally described by Ko filer and Milstein (1975); the human B-cell hybridoma technique (Cote et al, 1983); and the EBV-hybridoma technique (Cole et al. 1985). Alternatively, techniques described for the production of single chain antibodies (see e.g., U.S. Pat. No. 4,946,778) can be adapted to produce anti-CXCR3 single chain antibodies.
  • CXCR3 antagonists useful in practicing the present invention also include anti-CXCR3 antibody fragments including but not limited to F(ab') 2 fragments, which can be generated by pepsin digestion of an intact antibody molecule, and Fab fragments, which can be generated by reducing the disulfide bridges of the F(ab') 2 fragments.
  • Fab and/or scFv expression libraries can be constructed to allow rapid identification of fragments having the desired specificity to CXCR3.
  • Humanized anti-CXCR3 antibodies and antibody fragments therefrom can also be prepared according to known techniques.
  • “Humanized antibodies” are forms of non-human (e.g., rodent) chimeric antibodies that contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region (CDRs) of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity.
  • donor antibody such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity and capacity.
  • framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • humanized antibodies may comprise residues that are not found in the recipient antibody or in the donor antibody. These modifications are made to further refine antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence.
  • the humanized antibody optionally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • CXCR3 antagonists may be selected from aptamers.
  • Aptamers are a class of molecule that represents an alternative to antibodies in term of molecular recognition.
  • Aptamers are oligonucleotide or oligopeptide sequences with the capacity to recognize virtually any class of target molecules with high affinity and specificity.
  • Such ligands may be isolated through Systematic Evolution of Ligands by Exponential enrichment (SELEX) of a random sequence library, as described in Tuerk C. and Gold L., 1990.
  • the random sequence library is obtainable by combinatorial chemical synthesis of DNA. In this library, each member is a linear oligomer, eventually chemically modified, of a unique sequence. Possible modifications, uses and advantages of this class of molecules have been reviewed in Jayasena S.D., 1999.
  • Peptide aptamers consists of a conformationally constrained antibody variable region displayed by a platform protein, such as E. coli Thioredoxin A that are selected from combinatorial libraries by two hybrid methods (Colas et al, 1996).
  • the compound according to the invention is an inhibitor of the CXCR3 receptor expression.
  • Small inhibitory RNAs can also function as inhibitors of CXCR3 receptor gene expression for use in the present invention.
  • CXCR3 receptor gene expression can be reduced by contacting a subject or cell with a small double stranded RNA (dsRNA), or a vector or construct causing the production of a small double stranded RNA, such that CXCR3 receptor gene expression is specifically inhibited (i.e. RNA interference or RNAi).
  • dsRNA small double stranded RNA
  • RNAi RNA interference
  • Methods for selecting an appropriate dsRNA or dsRNA-encoding vector are well known in the art for genes whose sequence is known (e.g. see for example Tuschl, T. et al. (1999); Elbashir, S. M. et al. (2001); Hannon, GJ.
  • Ribozymes can also function as inhibitors of CXCR3 receptor gene expression for use in the present invention.
  • Ribozymes are enzymatic RNA molecules capable of catalyzing the specific cleavage of RNA.
  • the mechanism of ribozyme action involves sequence specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleo lytic cleavage.
  • Engineered hairpin or hammerhead motif ribozyme molecules that specifically and efficiently catalyze endonucleolytic cleavage of CXCR3 receptor mR A sequences are thereby useful within the scope of the present invention.
  • ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites, which typically include the following sequences, GUA, GUU, and GUC. Once identified, short RNA sequences of between about 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site can be evaluated for predicted structural features, such as secondary structure, that can render the oligonucleotide sequence unsuitable. The suitability of candidate targets can also be evaluated by testing their accessibility to hybridization with complementary oligonucleotides, using, e.g., ribonuclease protection assays.
  • antisense oligonucleotides and ribozymes useful as inhibitors of CXCR3 receptor gene expression can be prepared by known methods. These include techniques for chemical synthesis such as, e.g., by solid phase phosphoramadite chemical synthesis. Alternatively, anti-sense RNA molecules can be generated by in vitro or in vivo transcription of DNA sequences encoding the RNA molecule. Such DNA sequences can be incorporated into a wide variety of vectors that incorporate suitable RNA polymerase promoters such as the T7 or SP6 polymerase promoters. Various modifications to the oligonucleotides of the invention can be introduced as a means of increasing intracellular stability and half-life.
  • Possible modifications include but are not limited to the addition of flanking sequences of ribonucleotides or deoxyribonucleotides to the 5' and/or 3' ends of the molecule, or the use of phosphorothioate or 2'-0-methyl rather than phosphodiesterase linkages within the oligonucleotide backbone.
  • Antisense oligonucleotides siRNAs and ribozymes of the invention may be delivered in vivo alone or in association with a vector.
  • a "vector" is any vehicle capable of facilitating the transfer of the antisense oligonucleotide siRNA or ribozyme nucleic acid to the cells and preferably cells expressing CXCR3 receptor.
  • the vector transports the nucleic acid to cells with reduced degradation relative to the extent of degradation that would result in the absence of the vector.
  • the vectors useful in the invention include, but are not limited to, plasmids, phagemids, viruses, other vehicles derived from viral or bacterial sources that have been manipulated by the insertion or incorporation of the the antisense oligonucleotide siRNA or ribozyme nucleic acid sequences.
  • Viral vectors are a preferred type of vector and include, but are not limited to nucleic acid sequences from the following viruses: retrovirus, such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus; adenovirus, adeno-associated virus; SV40-type viruses; polyoma viruses; Epstein-Barr viruses; papilloma viruses; herpes virus; vaccinia virus; polio virus; and RNA virus such as a retrovirus.
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus
  • retrovirus such as moloney murine leukemia virus, harvey murine sarcoma virus, murine mammary tumor virus, and rouse sarcoma virus
  • adenovirus adeno
  • Non-cytopathic viruses include retroviruses (e.g., lentivirus), the life cycle of which involves reverse transcription of genomic viral RNA into DNA with subsequent proviral integration into host cellular DNA. Retroviruses have been approved for human gene therapy trials. Most useful are those retroviruses that are replication-deficient (i.e., capable of directing synthesis of the desired proteins, but incapable of manufacturing an infectious particle). Such genetically altered retroviral expression vectors have general utility for the high-efficiency transduction of genes in vivo.
  • adeno-viruses and adeno-associated viruses are double-stranded DNA viruses that have already been approved for human use in gene therapy.
  • the adeno-associated virus can be engineered to be replication deficient and is capable of infecting a wide range of cell types and species. It further has advantages such as, heat and lipid solvent stability; high transduction frequencies in cells of diverse lineages, including hemopoietic cells; and lack of superinfection inhibition thus allowing multiple series of transductions.
  • the adeno-associated virus can integrate into human cellular DNA in a site-specific manner, thereby minimizing the possibility of insertional mutagenesis and variability of inserted gene expression characteristic of retroviral infection.
  • adeno-associated virus infections have been followed in tissue culture for greater than 100 passages in the absence of selective pressure, implying that the adeno-associated virus genomic integration is a relatively stable event.
  • the adeno- associated virus can also function in an extrachromosomal fashion.
  • Plasmid vectors have been extensively described in the art and are well known to those of skill in the art. See e.g. Sambrook et al, 1989. In the last few years, plasmid vectors have been used as DNA vaccines for delivering antigen-encoding genes to cells in vivo. They are particularly advantageous for this because they do not have the same safety concerns as with many of the viral vectors. These plasmids, however, having a promoter compatible with the host cell, can express a peptide from a gene operatively encoded within the plasmid.
  • Plasmids may be delivered by a variety of parenteral, mucosal and topical routes.
  • the DNA plasmid can be injected by intramuscular, eye, intradermal, subcutaneous, or other routes. It may also be administered by intranasal sprays or drops, rectal suppository and orally.
  • the plasmids may be given in an aqueous solution, dried onto gold particles or in association with another DNA delivery system including but not limited to liposomes, dendrimers, cochleate and micro encap sulation.
  • the antisense oligonucleotide, siRNA, shRNA or ribozyme nucleic acid sequence is under the control of a heterologous regulatory region, e.g., a heterologous promoter.
  • the promoter may be specific for Muller glial cells, microglia cells, endothelial cells, pericyte cells and astrocytes
  • a specific expression in Muller glial cells may be obtained through the promoter of the glutamine synthetase gene is suitable.
  • the promoter can also be, e.g., a viral promoter, such as CMV promoter or any synthetic promoters.
  • Another object of the invention relates to a method for treating glaucoma comprising administering to a subject in need thereof a therapeutically effective amount of compound which is an antagonist of CXCR3 or an inhibitor of the CXCR3 receptor expression as described above.
  • the invention relates to a method for treating glaucoma comprising administering to a subject in need thereof a therapeutically effective amount of a CXCR3 antagonist as above described.
  • Compounds of the invention may be administered in the form of a pharmaceutical composition, as defined below.
  • said compound which is an antagonist of CXCR3 or an inhibitor of the CXCR3 receptor expression.
  • a “therapeutically effective amount” is meant a sufficient amount of compound to treat and/or to prevent glaucoma disorder.
  • the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed, the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific polypeptide employed; and like factors well known in the medical arts.
  • the daily dosage of the products may be varied over a wide range from 0.01 to 1,000 mg per adult per day.
  • the compositions contain 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 250 and 500 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • a medicament typically contains from about 0.01 mg to about 500 mg of the active ingredient, preferably from 1 mg to about 100 mg of the active ingredient.
  • An effective amount of the drug is ordinarily supplied at a dosage level from 0.0002 mg/kg to about 20 mg/kg of body weight per day, especially from about 0.001 mg/kg to 7 mg/kg of body weight per day.
  • Compounds according to the invention may be used for the preparation of a pharmaceutical composition for the treatment of a glaucoma disorder.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective dose of an antagonist of CXCR3 or an inhibitor of the CXCR3 receptor expression, preferably a CXCR3 antagonist, according to the invention.
  • Any therapeutic agent of the invention may be combined with pharmaceutically acceptable excipients, and optionally sustained-release matrices, such as biodegradable polymers, to form therapeutic compositions.
  • “Pharmaceutically” or “pharmaceutically acceptable” refers to molecular entities and compositions that do not produce an adverse, allergic or other untoward reaction when administered to a mammal, especially a human, as appropriate.
  • a pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • compositions for example, the route of administration, the dosage and the regimen naturally depend upon the condition to be treated, the severity of the illness, the age, weight, and sex of the patient, etc.
  • compositions of the invention can be formulated for a topical, oral, intranasal, parenteral, intraocular, intravenous, intramuscular or subcutaneous administration and the like.
  • compositions of the inventions may be formulated for intraocular either with eye drops, subconjunctival or intravitreal administration.
  • local ocular routes should be used such as intravitreous, topical, periocular injections (sub conjunctival, peri bulbar, latero bulbar, retro bulbar, sub tenon, supra choroidal), intra or peri ocular implants (intra sceral, peri scleral, episcleral), intra virteous implants, ocular surface implants or any releasing systems such as emulsions, solid non biodegradable or degradable implants or tablets, mini pumps or any topical formulations.
  • periocular injections sub conjunctival, peri bulbar, latero bulbar, retro bulbar, sub tenon, supra choroidal
  • intra or peri ocular implants intra sceral, peri scleral, episcleral
  • intra virteous implants ocular surface implants or any releasing systems such as emulsions, solid non biodegradable or degradable implants or tablets, mini pumps or any topical formulations.
  • the pharmaceutical compositions contain vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • vehicles which are pharmaceutically acceptable for a formulation capable of being injected.
  • These may be in particular isotonic, sterile, saline solutions (monosodium or disodium phosphate, sodium, potassium, calcium or magnesium chloride and the like or mixtures of such salts), or dry, especially freeze-dried compositions which upon addition, depending on the case, of sterilized water or physiological saline, permit the constitution of injectable solutions.
  • the doses used for the administration can be adapted as a function of various parameters, and in particular as a function of the mode of administration used, of the relevant pathology, or alternatively of the desired duration of treatment.
  • compositions include, e.g. tablets or other solids for oral administration; time release capsules; and any other form currently can be used.
  • Active ingredient may be also delivered directly to the eye by ocular tissue injection such as periocular, conjunctival, subtenon, intracameral, intravitreal, intraocular, subretinal, subconjunctival, retrobulbar, or intracanalicular injections; by direct application to the eye using a catheter or other placement device such as a retinal pellet, intraocular insert, suppository or an implant comprising a porous, non-porous, or gelatinous material; by topical ocular drops or ointments; or by a slow release device in the cul-de-sac or implanted adjacent to the sclera (transscleral) or in the sclera (intrascleral) or within the eye.
  • Intracameral injection may be through the cornea into the anterior chamber to allow the agent to reach the trabecular meshwork.
  • Intracanalicular injection may be into the venous collector channels draining Schlemm's canal or into Schlemm's canal
  • an active ingredient may be combined with ophthalmo logically acceptable preservatives, co-solvents, surfactants, viscosity enhancers, penetration enhancers, buffers, sodium chloride, or water to form an aqueous, sterile ophthalmic suspension or solution.
  • Solution formulations may be prepared by dissolving the active ingredient in a physiologically acceptable isotonic aqueous buffer. Further, the solution may include an acceptable surfactant to assist in dissolving the active ingredient.
  • Viscosity building agents such as hydro xymethyl cellulose, hydro xyethyl cellulose, methylcellulose, polyvinylpyrrolidone, or the like may be added to the compositions of the present invention to improve the retention of the compound.
  • a sterile ophthalmic ointment formulation the active ingredient is combined with a preservative in an appropriate vehicle, such as mineral oil, liquid lanolin, or white petrolatum.
  • an appropriate vehicle such as mineral oil, liquid lanolin, or white petrolatum.
  • Sterile ophthalmic gel formulations may be prepared by suspending the active ingredient in a hydrophilic base prepared from the combination of, for example,CARBOPOL®-940 (BF Goodrich, Charlotte, NC), or the like, according to methods known in the art.
  • VISCOAT® Alcon Laboratories, Inc., Fort Worth, TX
  • intraocular injection for example.
  • compositions of the present invention may contain penetration enhancing agents such as cremephor and TWEEN® 80 (polyoxy ethylene sorbitan monolaureate, Sigma Aldrich, St. Louis, MO), in the event the active ingredient is less penetrating in the eye.
  • penetration enhancing agents such as cremephor and TWEEN® 80 (polyoxy ethylene sorbitan monolaureate, Sigma Aldrich, St. Louis, MO), in the event the active ingredient is less penetrating in the eye.
  • FIGURE Figure 1 In vivo effect of the CXCR3 inhibitor NBI74330 on the intraocular pressure (IOP) in a rat model of surgically-induced glaucoma.
  • NBI74330 ⁇ or vehicle were performed in glaucoma eyes and in control eyes.
  • One single injection of NBI (A) induced a reduction in IOP during 3 days (p ⁇ 0.001).
  • Two injections of NBI (A and B) induced a decrease in IOP during 12 days (p ⁇ 0.001).
  • NBI or vehicle injections in control eyes did not influence the IOP. (In vivo measurements of the IOP using TonoPen).
  • FIG. 1 Effect of CXCR3 inhibitor NBI74330 on the intraocular pressure (IOP) in function of the dose.
  • Mann- Whitney U-test *, P ⁇ 0.05 and **, P ⁇ 0.01 relative to vehicle-treated eyes.
  • Figure 4 In vivo treatment with a CXCR3 antagonist increased aqueous humor outflow and trabecular filtration, and decreased trabecular cell apoptosis in an animal model of glaucoma.
  • Treatment with a CXCR3 antagonist lowers the intra ocular pressure and reduces TM cell apoptosis in a rat model of glaucoma.
  • TM was identified in each cryosections as a pigmented slim tissue on the posterior sclera-cornea beside the iris root and just above the endothelial Schlemm's canals.
  • Apoptosis was higher in trabecular cells of untreated glaucoma eyes than in glaucoma eyes treated with NBI74330.
  • endothelial Schlemm cells and retinal cells -especially in the inner nuclear layer and ganglion cell layer- presented more TUNEL labeling in the untreated glaucoma eyes than in glaucoma eyes treated with NBI74330.
  • NBI-74330 reduced IOP in a dose-dependent manner ( Figure 2).
  • a CXCR4 selective antagonist AMD- 3100
  • IOP IOP in glaucomatous eyes
  • NBI-74330 nor AMD-3100 (inhibitor of CXCR4) had any effect on IOP.
  • selective CXCR3 antagonist reduces IOP in a rat model of glaucoma, whereas selective CXCR4 antagonist has no effect.
  • CXCR3 receptor expression may be used for the treatment of glaucoma.
  • Aqueous humor outflow was measured by in vivo fluorophotometry ( Figure 4A) and the trabecular aqueous outflow evaluated by fluorescent microsphere injection and ex vivo confocal imaging of TMs (data not shown).
  • Figure 4A we observed a decrease in aqueous humor outflow along with a decrease in the TM filtrating surface in hypertensive eyes compared to controls 1 month after the surgical procedure.
  • This decrease in trabecular outflow facility is similar to what is observed during hypertensive POAG in humans.
  • aqueous humor outflow impairment was significantly reversed in NBI-74330-treated eyes compared to vehicle-treated controls.
  • rat TM tissues were assessed for cellularity and apoptosis by immunohistofluorescence and TUNEL-labeling.
  • NBI-related reduction in IOP was associated with a decrease in retinal ganglion cell apoptosis as assessed by TUNEL-labeling (4.01 ⁇ 0.86, 11.11 ⁇ 3.62, and 5.73 ⁇ 1.15 apoptotic retinal ganglion cells/mm for control eyes, untreated glaucomatous eyes, and NBI-treated glaucomatous eyes, respectively; P ⁇ 0.05 between each group).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Ophthalmology & Optometry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention a pour objet un composé qui est un antagoniste de CXCR3 ou un inhibiteur de l'expression du récepteur CXCR3 pour le traitement du glaucome.
EP11727194.0A 2010-06-28 2011-06-24 Composition pharmaceutique destinée à être utilisée dans le traitement du glaucome Withdrawn EP2585071A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP11727194.0A EP2585071A1 (fr) 2010-06-28 2011-06-24 Composition pharmaceutique destinée à être utilisée dans le traitement du glaucome

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP10305693 2010-06-28
PCT/EP2011/060638 WO2012000904A1 (fr) 2010-06-28 2011-06-24 Composition pharmaceutique destinée à être utilisée dans le traitement du glaucome
EP11727194.0A EP2585071A1 (fr) 2010-06-28 2011-06-24 Composition pharmaceutique destinée à être utilisée dans le traitement du glaucome

Publications (1)

Publication Number Publication Date
EP2585071A1 true EP2585071A1 (fr) 2013-05-01

Family

ID=42543252

Family Applications (1)

Application Number Title Priority Date Filing Date
EP11727194.0A Withdrawn EP2585071A1 (fr) 2010-06-28 2011-06-24 Composition pharmaceutique destinée à être utilisée dans le traitement du glaucome

Country Status (4)

Country Link
US (1) US9095591B2 (fr)
EP (1) EP2585071A1 (fr)
JP (1) JP5874124B2 (fr)
WO (1) WO2012000904A1 (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102126007B1 (ko) * 2010-01-22 2020-06-24 알러간, 인코포레이티드 전방내 서방성 치료제 이식물

Family Cites Families (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
AUPP249298A0 (en) 1998-03-20 1998-04-23 Ag-Gene Australia Limited Synthetic genes and genetic constructs comprising same I
GB9927444D0 (en) 1999-11-19 2000-01-19 Cancer Res Campaign Tech Inhibiting gene expression
CA2403397A1 (fr) 2000-03-16 2001-09-20 Genetica, Inc. Procedes et compositions d'interference d'arn
EP1554253A4 (fr) 2002-06-03 2006-09-20 Smithkline Beecham Corp Composes imidazolium inhibiteurs de cxcr3
US20040242498A1 (en) * 2003-02-27 2004-12-02 Collins Tassie L. CXCR3 antagonists
WO2005035534A1 (fr) * 2003-10-08 2005-04-21 Ono Pharmaceutical Co., Ltd. Composes a anneau bicyclique heterocyclique et a anneau tricyclique heterocyclique et medicaments les contenant
AU2006214477A1 (en) 2005-02-16 2006-08-24 Pharmacopeia, Inc. Heterocyclic substituted piperazines with CXCR3 antagonist activity
CA2598457A1 (fr) 2005-02-16 2006-08-24 Schering Corporation Piperazine-piperidines pyridyl et phenyl substituees a activite antagoniste des cxcr3
ATE518855T1 (de) 2005-02-16 2011-08-15 Schering Corp Neue heterozyklische substituierte pyridin- oder phenylverbindungen mit cxcr3-antagonistischer aktivität
CN101203509B (zh) 2005-02-16 2013-05-08 默沙东公司 具有cxcr3拮抗剂活性的胺-连接的吡啶基和苯基取代的哌嗪-哌啶
MX2007010068A (es) 2005-02-16 2007-10-10 Schering Corp Piperazino-piperidinas con actividad antagonista de cxcr3.
MX2007009947A (es) 2005-02-16 2007-09-26 Schering Corp Piperazin-piperidinas sustituidas con pirazinilo con actividad antagonista de cxcr3.
MX2007009949A (es) 2005-02-16 2007-09-26 Schering Corp Pirazinil-piperazin-piperidinas sustituidas con heteroarilo con actividad antagonista de cxcr3.
EP1885386A4 (fr) * 2005-05-18 2009-01-07 Intermune Inc Ligands recepteur de chimiokine non naturelle et procedes d'utilisation correspondants
GB0511060D0 (en) 2005-05-31 2005-07-06 Novartis Ag Organic compounds
US7781437B2 (en) 2005-10-11 2010-08-24 Schering Corporation Substituted heterocyclic compounds with CXCR3 antagonist activity
WO2007064553A2 (fr) 2005-11-29 2007-06-07 Merck & Co., Inc. Derives de thiazole comme modulateurs de recepteurs cxcr3
CN101460482B (zh) 2006-03-21 2013-03-27 默沙东公司 具有cxcr3拮抗剂活性的杂环取代的吡啶化合物
CN101516869A (zh) 2006-07-14 2009-08-26 先灵公司 具有cxcr3拮抗剂活性的杂环取代的哌嗪化合物
EP2187900B1 (fr) * 2007-08-16 2016-11-09 The Schepens Eye Research Institute, Inc. Composition thérapeutique pour le traitement de l'inflammation des annexes et des tissus oculaires

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
None *
See also references of WO2012000904A1 *

Also Published As

Publication number Publication date
US9095591B2 (en) 2015-08-04
JP2013529661A (ja) 2013-07-22
US20130172370A1 (en) 2013-07-04
WO2012000904A1 (fr) 2012-01-05
JP5874124B2 (ja) 2016-03-02

Similar Documents

Publication Publication Date Title
EP1993584B1 (fr) Inhibiteurs de l'activité de CXCR4 pour son utilisation dans le traitement des maladies oculaires
JP4951339B2 (ja) 眼血管新生疾患の治療のための併用療法
AU2015257651A1 (en) Compounds for treating ophthalmic diseases and disorders
US20090098139A1 (en) Combination therapy for the treatment of neovascular disorders
EA022873B1 (ru) Применение ингибитора vegf-r2 для лечения офтальмологического сосудистого заболевания
US9155790B2 (en) Methods and compositions for modulating ocular damage
EP2671589A1 (fr) Agent médicinal pour la prévention ou le traitement de maladies associées à une néovascularisation intraoculaire et/ou une hyperperméabilité vasculaire intraoculaire
US9095591B2 (en) Pharmaceutical composition for use in the treatment of glaucoma
JP2015500221A (ja) 眼疾患の治療および予防方法
WO2014139014A1 (fr) Méthodes et compositions pour l'inhibition de l'activité du facteur de croissance de l'endothélium vasculaire et de la perméabilité vasculaire
CA3204497A1 (fr) Procedes et compositions utilisant des cellules de vascularisation dependantes de l'ido1 pour le traitement d'etats pathologiques impliquant une neovascularisation
WO2017091473A1 (fr) Composition de blocage de l'angiogenèse
JP2014511392A (ja) 創傷を治癒または治療するための分子標的
CN112888440A (zh) Akt抑制剂在眼科中的用途
CA3037116A1 (fr) Methodes de traitement d'une maladie oculaire a l'aide d'inhibiteurs de csf-1r
US20230077811A1 (en) Activation of neuropeptide receptors on plasmacytoid dendritic cells to treat or prevent ocular diseases associated with neovascularization and inflammation
US20160304881A1 (en) Ddr1 antagonist or an inhibitor of ddr1 gene expression for use in the prevention or treatment of crescentic glomerulonephritis

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20130122

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20170804

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20171215